Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Med ; 30(7): 1913-1922, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38844796

RESUMO

Recent research showed that precision medicine can identify new treatment strategies for patients with childhood cancers. However, it is unclear which patients will benefit most from precision-guided treatment (PGT). Here we report consecutive data from 384 patients with high-risk pediatric cancer (with an expected cure rate of less than 30%) who had at least 18 months of follow-up on the ZERO Childhood Cancer Precision Medicine Program PRecISion Medicine for Children with Cancer (PRISM) trial. A total of 256 (67%) patients received PGT recommendations and 110 (29%) received a recommended treatment. PGT resulted in a 36% objective response rate and improved 2-year progression-free survival compared with standard of care (26% versus 12%; P = 0.049) or targeted agents not guided by molecular findings (26% versus 5.2%; P = 0.003). PGT based on tier 1 evidence, PGT targeting fusions or commenced before disease progression had the greatest clinical benefit. Our data show that PGT informed by comprehensive molecular profiling significantly improves outcomes for children with high-risk cancers. ClinicalTrials.gov registration: NCT03336931.


Assuntos
Neoplasias , Medicina de Precisão , Humanos , Medicina de Precisão/métodos , Criança , Neoplasias/genética , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Feminino , Masculino , Adolescente , Pré-Escolar , Lactente , Intervalo Livre de Progressão , Resultado do Tratamento
2.
Cancer Res ; 83(16): 2716-2732, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37523146

RESUMO

For one-third of patients with pediatric cancer enrolled in precision medicine programs, molecular profiling does not result in a therapeutic recommendation. To identify potential strategies for treating these high-risk pediatric patients, we performed in vitro screening of 125 patient-derived samples against a library of 126 anticancer drugs. Tumor cell expansion did not influence drug responses, and 82% of the screens on expanded tumor cells were completed while the patients were still under clinical care. High-throughput drug screening (HTS) confirmed known associations between activating genomic alterations in NTRK, BRAF, and ALK and responses to matching targeted drugs. The in vitro results were further validated in patient-derived xenograft models in vivo and were consistent with clinical responses in treated patients. In addition, effective combinations could be predicted by correlating sensitivity profiles between drugs. Furthermore, molecular integration with HTS identified biomarkers of sensitivity to WEE1 and MEK inhibition. Incorporating HTS into precision medicine programs is a powerful tool to accelerate the improved identification of effective biomarker-driven therapeutic strategies for treating high-risk pediatric cancers. SIGNIFICANCE: Integrating HTS with molecular profiling is a powerful tool for expanding precision medicine to support drug treatment recommendations and broaden the therapeutic options available to high-risk pediatric cancers.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Criança , Avaliação Pré-Clínica de Medicamentos , Detecção Precoce de Câncer , Neoplasias/tratamento farmacológico , Neoplasias/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ensaios de Triagem em Larga Escala/métodos
3.
Genome Med ; 15(1): 20, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37013636

RESUMO

BACKGROUND: Molecular profiling of the tumour immune microenvironment (TIME) has enabled the rational choice of immunotherapies in some adult cancers. In contrast, the TIME of paediatric cancers is relatively unexplored. We speculated that a more refined appreciation of the TIME in childhood cancers, rather than a reliance on commonly used biomarkers such as tumour mutation burden (TMB), neoantigen load and PD-L1 expression, is an essential prerequisite for improved immunotherapies in childhood solid cancers. METHODS: We combined immunohistochemistry (IHC) with RNA sequencing and whole-genome sequencing across a diverse spectrum of high-risk paediatric cancers to develop an alternative, expression-based signature associated with CD8+ T-cell infiltration of the TIME. Furthermore, we explored transcriptional features of immune archetypes and T-cell receptor sequencing diversity, assessed the relationship between CD8+ and CD4+ abundance by IHC and deconvolution predictions and assessed the common adult biomarkers such as neoantigen load and TMB. RESULTS: A novel 15-gene immune signature, Immune Paediatric Signature Score (IPASS), was identified. Using this signature, we estimate up to 31% of high-risk cancers harbour infiltrating T-cells. In addition, we showed that PD-L1 protein expression is poorly correlated with PD-L1 RNA expression and TMB and neoantigen load are not predictive of T-cell infiltration in paediatrics. Furthermore, deconvolution algorithms are only weakly correlated with IHC measurements of T-cells. CONCLUSIONS: Our data provides new insights into the variable immune-suppressive mechanisms dampening responses in paediatric solid cancers. Effective immune-based interventions in high-risk paediatric cancer will require individualised analysis of the TIME.


Assuntos
Antígeno B7-H1 , Neoplasias , Adulto , Humanos , Criança , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias/genética , Linfócitos T CD8-Positivos/metabolismo , Biomarcadores Tumorais/genética , Microambiente Tumoral/genética , Mutação
4.
Front Oncol ; 13: 1123492, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36937401

RESUMO

Introduction: Ependymomas (EPN) are the third most common malignant brain cancer in children. Treatment strategies for pediatric EPN have remained unchanged over recent decades, with 10-year survival rates stagnating at just 67% for children aged 0-14 years. Moreover, a proportion of patients who survive treatment often suffer long-term neurological side effects as a result of therapy. It is evident that there is a need for safer, more effective treatments for pediatric EPN patients. There are ten distinct subgroups of EPN, each with their own molecular and prognostic features. To identify and facilitate the testing of new treatments for EPN, in vivo laboratory models representative of the diverse molecular subtypes are required. Here, we describe the establishment of a patient-derived orthotopic xenograft (PDOX) model of posterior fossa A (PFA) EPN, derived from a metastatic cranial lesion. Methods: Patient and PDOX tumors were analyzed using immunohistochemistry, DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. Results: Both patient and PDOX tumors classified as PFA EPN by methylation profiling, and shared similar histological features consistent with this molecular subgroup. RNA sequencing revealed that gene expression patterns were maintained across the primary and metastatic tumors, as well as the PDOX. Copy number profiling revealed gains of chromosomes 7, 8 and 19, and loss of chromosomes 2q and 6q in the PDOX and matched patient tumor. No clinically significant single nucleotide variants were identified, consistent with the low mutation rates observed in PFA EPN. Overexpression of EZHIP RNA and protein, a common feature of PFA EPN, was also observed. Despite the aggressive nature of the tumor in the patient, this PDOX was unable to be maintained past two passages in vivo. Discussion: Others who have successfully developed PDOX models report some of the lowest success rates for EPN compared to other pediatric brain cancer types attempted, with loss of tumorigenicity not uncommon, highlighting the challenges of propagating these tumors in the laboratory. Here, we discuss our collective experiences with PFA EPN PDOX model generation and propose potential approaches to improve future success in establishing preclinical EPN models.

5.
Sci Rep ; 13(1): 3775, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36882456

RESUMO

Diffuse midline gliomas (DMG) harbouring H3K27M mutation are paediatric tumours with a dismal outcome. Recently, a new subtype of midline gliomas has been described with similar features to DMG, including loss of H3K27 trimethylation, but lacking the canonical H3K27M mutation (H3-WT). Here, we report a cohort of five H3-WT tumours profiled by whole-genome sequencing, RNA sequencing and DNA methylation profiling and combine their analysis with previously published cases. We show that these tumours have recurrent and mutually exclusive mutations in either ACVR1 or EGFR and are characterised by high expression of EZHIP associated to its promoter hypomethylation. Affected patients share a similar poor prognosis as patients with H3K27M DMG. Global molecular analysis of H3-WT and H3K27M DMG reveal distinct transcriptome and methylome profiles including differential methylation of homeobox genes involved in development and cellular differentiation. Patients have distinct clinical features, with a trend demonstrating ACVR1 mutations occurring in H3-WT tumours at an older age. This in-depth exploration of H3-WT tumours further characterises this novel DMG, H3K27-altered sub-group, characterised by a specific immunohistochemistry profile with H3K27me3 loss, wild-type H3K27M and positive EZHIP. It also gives new insights into the possible mechanism and pathway regulation in these tumours, potentially opening new therapeutic avenues for these tumours which have no known effective treatment. This study has been retrospectively registered on clinicaltrial.gov on 8 November 2017 under the registration number NCT03336931 ( https://clinicaltrials.gov/ct2/show/NCT03336931 ).


Assuntos
Genes Homeobox , Glioma , Criança , Humanos , Histonas/genética , Metilação , Glioma/genética , Mutação , Receptores ErbB/genética , Receptores de Ativinas Tipo I
6.
Br J Cancer ; 127(5): 908-915, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35650277

RESUMO

BACKGROUND: ABL-class fusions including NUP214-ABL1 and EBF1-PDGFRB occur in high risk acute lymphoblastic leukaemia (ALL) with gene expression patterns similar to BCR-ABL-positive ALL. Our aim was to evaluate new DNA-based measurable residual disease (MRD) tests detecting these fusions and IKZF1-deletions in comparison with conventional immunoglobulin/T-cell receptor (Ig/TCR) markers. METHODS: Precise genomic breakpoints were defined from targeted or whole genome next generation sequencing for ABL-fusions and BCR-ABL1. Quantitative PCR assays were designed and used to re-measure MRD in remission bone marrow samples previously tested using Ig/TCR markers. All MRD testing complied with EuroMRD guidelines. RESULTS: ABL-class patients had 46% 5year event-free survival and 79% 5year overall survival. All had sensitive fusion tests giving high concordance between Ig/TCR and ABL-class fusion results (21 patients, n = 257 samples, r2 = 0.9786, P < 0.0001) and Ig/TCR and IKZF1-deletion results (9 patients, n = 143 samples, r2 = 0.9661, P < 0.0001). In contrast, in BCR-ABL1 patients, Ig/TCR and BCR-ABL1 tests were discordant in 32% (40 patients, n = 346 samples, r2 = 0.4703, P < 0.0001) and IKZF1-deletion results were closer to Ig/TCR (25 patients, n = 176, r2 = 0.8631, P < 0.0001). CONCLUSIONS: MRD monitoring based on patient-specific assays detecting gene fusions or recurrent assays for IKZF1-deletions is feasible and provides good alternatives to Ig/TCR tests to monitor MRD in ABL-class ALL.


Assuntos
Proteínas de Fusão bcr-abl , Leucemia-Linfoma Linfoblástico de Células Precursoras , Criança , Proteínas de Fusão bcr-abl/genética , Humanos , Imunoglobulinas , Neoplasia Residual/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Antígenos de Linfócitos T/genética
7.
EMBO Mol Med ; 14(4): e14608, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-34927798

RESUMO

Biomarkers which better match anticancer drugs with cancer driver genes hold the promise of improved clinical responses and cure rates. We developed a precision medicine platform of rapid high-throughput drug screening (HTS) and patient-derived xenografting (PDX) of primary tumor tissue, and evaluated its potential for treatment identification among 56 consecutively enrolled high-risk pediatric cancer patients, compared with conventional molecular genomics and transcriptomics. Drug hits were seen in the majority of HTS and PDX screens, which identified therapeutic options for 10 patients for whom no targetable molecular lesions could be found. Screens also provided orthogonal proof of drug efficacy suggested by molecular analyses and negative results for some molecular findings. We identified treatment options across the whole testing platform for 70% of patients. Only molecular therapeutic recommendations were provided to treating oncologists and led to a change in therapy in 53% of patients, of whom 29% had clinical benefit. These data indicate that in vitro and in vivo drug screening of tumor cells could increase therapeutic options and improve clinical outcomes for high-risk pediatric cancer patients.


Assuntos
Antineoplásicos , Neoplasias , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Criança , Modelos Animais de Doenças , Genômica/métodos , Humanos , Neoplasias/patologia , Medicina de Precisão/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cell Genom ; 2(4): 100112, 2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-36776527

RESUMO

Complex somatic genomic rearrangements and copy number alterations are hallmarks of nearly all cancers. We have developed an algorithm, LINX, to aid interpretation of structural variant and copy number data derived from short-read, whole-genome sequencing. LINX classifies raw structural variant calls into distinct events and predicts their effect on the local structure of the derivative chromosome and the functional impact on affected genes. Visualizations facilitate further investigation of complex rearrangements. LINX allows insights into a diverse range of structural variation events and can reliably detect pathogenic rearrangements, including gene fusions, immunoglobulin enhancer rearrangements, intragenic deletions, and duplications. Uniquely, LINX also predicts chained fusions that we demonstrate account for 13% of clinically relevant oncogenic fusions. LINX also reports a class of inactivation events that we term homozygous disruptions that may be a driver mutation in up to 9% of tumors and may frequently affect PTEN, TP53, and RB1.

9.
Br J Cancer ; 126(3): 482-491, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34471258

RESUMO

BACKGROUND: Minimal residual disease (MRD) measurement is a cornerstone of contemporary acute lymphoblastic leukaemia (ALL) treatment. The presence of immunoglobulin (Ig) and T cell receptor (TCR) gene recombinations in leukaemic clones allows widespread use of patient-specific, DNA-based MRD assays. In contrast, paediatric solid tumour MRD remains experimental and has focussed on generic assays targeting tumour-specific messenger RNA, methylated DNA or microRNA. METHODS: We examined the feasibility of using whole-genome sequencing (WGS) data to design tumour-specific polymerase chain reaction (PCR)-based MRD tests (WGS-MRD) in 18 children with high-risk relapsed cancer, including ALL, high-risk neuroblastoma (HR-NB) and Ewing sarcoma (EWS) (n = 6 each). RESULTS: Sensitive WGS-MRD assays were generated for each patient and allowed quantitation of 1 tumour cell per 10-4 (0.01%)-10-5 (0.001%) mononuclear cells. In ALL, WGS-MRD and Ig/TCR-MRD were highly concordant. WGS-MRD assays also showed good concordance between quantitative PCR and droplet digital PCR formats. In serial clinical samples, WGS-MRD correlated with disease course. In solid tumours, WGS-MRD assays were more sensitive than RNA-MRD assays. CONCLUSIONS: WGS facilitated the development of patient-specific MRD tests in ALL, HR-NB and EWS with potential clinical utility in monitoring treatment response. WGS data could be used to design patient-specific MRD assays in a broad range of tumours.


Assuntos
Biomarcadores Tumorais/genética , Rearranjo Gênico , Neoplasia Residual/patologia , Neuroblastoma/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Sarcoma de Ewing/patologia , Sequenciamento Completo do Genoma/métodos , Adolescente , Neoplasias Ósseas/sangue , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Proteína Proto-Oncogênica N-Myc/genética , Neoplasia Residual/genética , Neuroblastoma/sangue , Neuroblastoma/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteína Proto-Oncogênica c-fli-1/genética , Receptores de Antígenos de Linfócitos T/genética , Sarcoma de Ewing/sangue , Sarcoma de Ewing/genética , Regulador Transcricional ERG/genética
10.
Acta Neuropathol Commun ; 9(1): 147, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493325

RESUMO

Diffuse leptomeningeal glioneuronal tumours (DLGNT) represent rare enigmatic CNS tumours of childhood. Most patients with this disease share common radiological and histopathological features but the clinical course of this disease is variable. A radiological hallmark of this disease is widespread leptomeningeal enhancement that may involve the entire neuroaxis with predilection for the posterior fossa and spine. The classic pathologic features include low- to moderate-density cellular lesions with OLIG2 expression and evidence of 'oligodendroglioma-like' appearance. The MAPK/ERK signaling pathway has recently been reported as a potential driver of tumourigenesis in up to 80% of DLGNT with KIAA1549:BRAF fusions being the most common event seen. Until now, limited analysis of the biological drivers of tumourigenesis has been undertaken via targeted profiling, chromosomal analysis and immunohistochemistry. Our study represents the first examples of comprehensive genomic sequencing in DLGNT and shows that it is not only feasible but crucial to our understanding of this rare disease. Moreover, we demonstrate that DLGNT may be more genomically complex than single-event MAPK/ERK signaling pathway tumours.


Assuntos
Neoplasias Encefálicas/genética , Genômica/métodos , Neoplasias Meníngeas/genética , Meningioma/genética , Neoplasias da Medula Espinal/genética , Adolescente , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamento farmacológico , Criança , Feminino , Humanos , Masculino , Neoplasias Meníngeas/diagnóstico , Neoplasias Meníngeas/tratamento farmacológico , Meningioma/diagnóstico , Meningioma/tratamento farmacológico , Neoplasias da Medula Espinal/diagnóstico , Neoplasias da Medula Espinal/tratamento farmacológico
11.
NPJ Precis Oncol ; 5(1): 9, 2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33580196

RESUMO

The prognosis of recurrent malignant peripheral nerve sheath tumors (MPNST) is dismal, with surgical resection being the only definitive salvage therapy. Treatment with chemoradiation approaches has not significantly improved patient outcomes. Similarly, trials of therapies targeting MPNST genomic drivers have thus far been unsuccessful. Improved understanding of the molecular pathogenesis of MPNST indicates frequent activation of the mitogen-activated protein kinase (MAPK) cell signaling pathway. MEK inhibitors have shown activity in preclinical studies; however, their clinical efficacy has not been reported to date. We describe here a case of sustained complete response to MEK inhibition in an adolescent patient with a recurrent metastatic MPNST with multiple alterations in the MAPK pathway, guided by a precision oncology approach.

12.
Clin Cancer Res ; 27(5): 1438-1451, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33310889

RESUMO

PURPOSE: TERT gene rearrangement with transcriptional superenhancers leads to TERT overexpression and neuroblastoma. No targeted therapy is available for clinical trials in patients with TERT-rearranged neuroblastoma. EXPERIMENTAL DESIGN: Anticancer agents exerting the best synergistic anticancer effects with BET bromodomain inhibitors were identified by screening an FDA-approved oncology drug library. The synergistic effects of the BET bromodomain inhibitor OTX015 and the proteasome inhibitor carfilzomib were examined by immunoblot and flow cytometry analysis. The anticancer efficacy of OTX015 and carfilzomib combination therapy was investigated in mice xenografted with TERT-rearranged neuroblastoma cell lines or patient-derived xenograft (PDX) tumor cells, and the role of TERT reduction in the anticancer efficacy was examined through rescue experiments in mice. RESULTS: The BET bromodomain protein BRD4 promoted TERT-rearranged neuroblastoma cell proliferation through upregulating TERT expression. Screening of an approved oncology drug library identified the proteasome inhibitor carfilzomib as the agent exerting the best synergistic anticancer effects with BET bromodomain inhibitors including OTX015. OTX015 and carfilzomib synergistically reduced TERT protein expression, induced endoplasmic reticulum stress, and induced TERT-rearranged neuroblastoma cell apoptosis which was blocked by TERT overexpression and endoplasmic reticulum stress antagonists. In mice xenografted with TERT-rearranged neuroblastoma cell lines or PDX tumor cells, OTX015 and carfilzomib synergistically blocked TERT expression, induced tumor cell apoptosis, suppressed tumor progression, and improved mouse survival, which was largely reversed by forced TERT overexpression. CONCLUSIONS: OTX015 and carfilzomib combination therapy is likely to be translated into the first clinical trial of a targeted therapy in patients with TERT-rearranged neuroblastoma.


Assuntos
Acetanilidas/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Rearranjo Gênico , Compostos Heterocíclicos com 3 Anéis/farmacologia , Terapia de Alvo Molecular/métodos , Neuroblastoma/tratamento farmacológico , Oligopeptídeos/farmacologia , Telomerase/genética , Fatores de Transcrição/antagonistas & inibidores , Animais , Apoptose , Proliferação de Células , Quimioterapia Combinada , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Inibidores de Proteassoma/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Artigo em Inglês | MEDLINE | ID: mdl-33144287

RESUMO

The identification of rearrangements driving expression of neurotrophic receptor tyrosine kinase (NTRK) family kinases in tumors has become critically important because of the availability of effective, specific inhibitor drugs. Whole-genome sequencing (WGS) combined with RNA sequencing (RNA-seq) can identify novel and recurrent expressed fusions. Here we describe three SPECC1L-NTRK fusions identified in two pediatric central nervous system cancers and an extracranial solid tumor using WGS and RNA-seq. These fusions arose either through a simple balanced rearrangement or in the context of a complex chromoplexy event. We cloned the SPECC1L-NTRK2 fusion directly from a patient sample and showed that enforced expression of this fusion is sufficient to promote cytokine-independent survival and proliferation. Cells transformed by SPECC1L-NTRK2 expression are sensitive to a TRK inhibitor drug. We report here that SPECC1L-NTRK fusions can arise in a range of pediatric cancers. Although WGS and RNA-seq are not required to detect NTRK fusions, these techniques may be of benefit when NTRK fusions are not suspected on clinical grounds or not identified by other methods.


Assuntos
Neoplasias Encefálicas/genética , Glicoproteínas de Membrana/genética , Proteínas de Fusão Oncogênica/genética , Fosfoproteínas/genética , Receptor trkA/genética , Receptor trkB/genética , Sarcoma/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Neoplasias do Sistema Nervoso Central/genética , Criança , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Masculino , Inibidores de Proteínas Quinases , Sarcoma/patologia , Sequenciamento Completo do Genoma
14.
Cancers (Basel) ; 12(10)2020 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-33053751

RESUMO

Radiation-induced glioma (RIG) is a highly aggressive brain cancer arising as a consequence of radiation therapy. We report a case of RIG that arose in the brain stem following treatment for paediatric medulloblastoma, and the development and characterisation of a matched orthotopic patient-derived xenograft (PDX) model (TK-RIG915). Patient and PDX tumours were analysed using DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. While initially thought to be a diffuse intrinsic pontine glioma (DIPG) based on disease location, results from methylation profiling and WGS were not consistent with this diagnosis. Furthermore, clustering analyses based on RNA expression suggested the tumours were distinct from primary DIPG. Additional gene expression analysis demonstrated concordance with a published RIG expression profile. Multiple genetic alterations that enhance PI3K/AKT and Ras/Raf/MEK/ERK signalling were discovered in TK-RIG915 including an activating mutation in PIK3CA, upregulation of PDGFRA and AKT2, inactivating mutations in NF1, and a gain-of-function mutation in PTPN11. Additionally, deletion of CDKN2A/B, increased IDH1 expression, and decreased ARID1A expression were observed. Detection of phosphorylated S6, 4EBP1 and ERK via immunohistochemistry confirmed PI3K pathway and ERK activation. Here, we report one of the first PDX models for RIG, which recapitulates the patient disease and is molecularly distinct from primary brain stem glioma. Genetic interrogation of this model has enabled the identification of potential therapeutic vulnerabilities in this currently incurable disease.

15.
Nat Med ; 26(11): 1742-1753, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33020650

RESUMO

The Zero Childhood Cancer Program is a precision medicine program to benefit children with poor-outcome, rare, relapsed or refractory cancer. Using tumor and germline whole genome sequencing (WGS) and RNA sequencing (RNAseq) across 252 tumors from high-risk pediatric patients with cancer, we identified 968 reportable molecular aberrations (39.9% in WGS and RNAseq, 35.1% in WGS only and 25.0% in RNAseq only). Of these patients, 93.7% had at least one germline or somatic aberration, 71.4% had therapeutic targets and 5.2% had a change in diagnosis. WGS identified pathogenic cancer-predisposing variants in 16.2% of patients. In 76 central nervous system tumors, methylome analysis confirmed diagnosis in 71.1% of patients and contributed to a change of diagnosis in two patients (2.6%). To date, 43 patients have received a recommended therapy, 38 of whom could be evaluated, with 31% showing objective evidence of clinical benefit. Comprehensive molecular profiling resolved the molecular basis of virtually all high-risk cancers, leading to clinical benefit in some patients.


Assuntos
Epigenoma/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Transcriptoma/genética , Adolescente , Criança , Pré-Escolar , Metilação de DNA/genética , Feminino , Humanos , Lactente , Masculino , Mutação/genética , Neoplasias/classificação , Neoplasias/patologia , Pediatria , Medicina de Precisão , Fatores de Risco , Sequenciamento do Exoma , Sequenciamento Completo do Genoma
16.
BMC Mol Cell Biol ; 21(1): 26, 2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32293262

RESUMO

BACKGROUND: Progesterone receptor membrane component 1 (PGRMC1) is often elevated in cancers, and exists in alternative states of phosphorylation. A motif centered on PGRMC1 Y180 was evolutionarily acquired concurrently with the embryological gastrulation organizer that orchestrates vertebrate tissue differentiation. RESULTS: Here, we show that mutagenic manipulation of PGRMC1 phosphorylation alters cell metabolism, genomic stability, and CpG methylation. Each of several mutants elicited distinct patterns of genomic CpG methylation. Mutation of S57A/Y180/S181A led to increased net hypermethylation, reminiscent of embryonic stem cells. Pathways enrichment analysis suggested modulation of processes related to animal cell differentiation status and tissue identity, as well as cell cycle control and ATM/ATR DNA damage repair regulation. We detected different genomic mutation rates in culture. CONCLUSIONS: A companion manuscript shows that these cell states dramatically affect protein abundances, cell and mitochondrial morphology, and glycolytic metabolism. We propose that PGRMC1 phosphorylation status modulates cellular plasticity mechanisms relevant to early embryological tissue differentiation.


Assuntos
Fosforilação , Receptores de Progesterona , Animais , Diferenciação Celular , Linhagem Celular , Metilação de DNA , Doença , Embriologia , Epigenômica , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Taxa de Mutação , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/biossíntese , Receptores de Progesterona/metabolismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-30936196

RESUMO

Adrenocortical carcinoma is a rare malignancy with a poor prognosis and few treatment options. Molecular characterization of this cancer remains limited. We present a case of an adrenocortical carcinoma (ACC) in a 37-yr-old female, with dual lung metastases identified 1 yr following commencement of adjuvant mitotane therapy. As standard therapeutic regimens are often unsuccessful in ACC, we undertook a comprehensive genomic study into this case to identify treatment options and monitor disease progress. We performed targeted and whole-genome sequencing of germline, primary tumor, and both metastatic tumors from this patient and monitored recurrence over 2 years using liquid biopsy for ctDNA and steroid hormone measurements. Sequencing revealed the primary and metastatic tumors were hyperhaploid, with extensive loss of heterozygosity but few structural rearrangements. Loss-of-function mutations were identified in MSH2, TP53, RB1, and PTEN, resulting in tumors with mismatch repair signatures and microsatellite instability. At the cellular level, tumors were populated by mitochondria-rich oncocytes. Longitudinal ctDNA mutation and hormone profiles were unable to detect micrometastatic disease, consistent with clinical indicators of disease remission. The molecular signatures in our ACC case suggested immunotherapy in the event of disease progression; however, the patient remains free of cancer. The extensive molecular analysis presented here could be applied to other rare and/or poorly stratified cancers to identify novel or repurpose existing therapeutic options, thereby broadly improving diagnoses, treatments, and prognoses.


Assuntos
Neoplasias do Córtex Suprarrenal/diagnóstico , Carcinoma Adrenocortical/diagnóstico , Neoplasias Pulmonares/secundário , Sequenciamento Completo do Genoma/métodos , Neoplasias do Córtex Suprarrenal/genética , Carcinoma Adrenocortical/genética , Adulto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Biópsia Líquida , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Instabilidade de Microssatélites , Mutação , Prognóstico
18.
Br J Cancer ; 119(6): 693-696, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30220707

RESUMO

Genes encoding TRK are oncogenic drivers in multiple tumour types including infantile fibrosarcoma, papillary thyroid cancer and high-grade gliomas (HGG). TRK fusions have a critical role in tumourigenesis in 40% of infant HGG. Here we report the first case of a TRK fusion-driven HGG treated with larotrectinib-the first selective pan-TRK inhibitor in clinical development. This 3-year-old girl had failed multiple therapies including chemotherapy and radiotherapy. Tumour profiling confirmed an ETV6-NTRK3 fusion. Treatment with larotrectinib led to rapid clinical improvement with near total resolution of primary and metastatic lesions on MRI imaging. This is the first report of a TRK fusion glioma successfully treated with a TRK inhibitor.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Pré-Escolar , Feminino , Glioma/diagnóstico por imagem , Glioma/genética , Glioma/patologia , Humanos , Imageamento por Ressonância Magnética , Gradação de Tumores , Proteínas de Fusão Oncogênica/genética , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Resultado do Tratamento , Sequenciamento Completo do Genoma
19.
PLoS One ; 7(10): e48591, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23119064

RESUMO

Inhibitors of DNA binding and differentiation (ID) proteins, a dominant-negative group of helix-loop-helix (HLH) transcription regulators, are well-characterized key players in cellular fate determination during development in mammals as well as Drosophila. Although not oncogenes themselves, their upregulation by various oncogenic proteins (such as Ras, Myc) and their inhibitory effects on cell cycle proteins (such as pRb) hint at their possible roles in tumorigenesis. Furthermore, their potency as inhibitors of cellular differentiation, through their heterodimerization with subsequent inactivation of the ubiquitous E proteins, suggest possible novel roles in engineering induced pluripotent stem cells (iPSCs). We present the high-resolution 2.1Å crystal structure of ID2 (HLH domain), coupled with novel biochemical insights in the presence of a divalent ion, possibly calcium (Ca2+), in the loop of ID proteins, which appear to be crucial for the structure and activity of ID proteins. These new insights will pave the way for new rational drug designs, in addition to current synthetic peptide options, against this potent player in tumorigenesis as well as more efficient ways for stem cells reprogramming.


Assuntos
Cátions Bivalentes/química , Proteína 2 Inibidora de Diferenciação/química , Proteínas Inibidoras de Diferenciação/química , Proteínas de Neoplasias/química , Multimerização Proteica , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Sítios de Ligação/genética , Cálcio/química , Cálcio/metabolismo , Cátions Bivalentes/metabolismo , Cristalografia por Raios X , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/genética , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Proteínas Inibidoras de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteína MyoD/química , Proteína MyoD/genética , Proteína MyoD/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Fator 3 de Transcrição/química , Fator 3 de Transcrição/genética , Fator 3 de Transcrição/metabolismo
20.
Br J Nutr ; 106(8): 1231-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21736825

RESUMO

Salmon provides long-chain (LC) n-3 PUFA and Se, which are well recognised for their health benefits. The n-3 and Se status of the New Zealand population is marginal. The objective of the present study was to compare the effects of consuming salmon v. supplementation with salmon oil on LC n-3 and Se status. Healthy volunteers (n 44) were randomly assigned to one of four groups consuming 2 × 120 g servings of salmon/week or 2, 4 or 6 salmon oil capsules/d for 8 weeks. Linear regression analysis predictive models were fitted to the capsule data to predict changes in erythrocyte LC n-3 levels with intakes of LC n-3 from capsules in amounts equivalent to that consumed from salmon. Changes in Se status (plasma Se and whole-blood glutathione peroxidase) were compared between the groups consuming salmon and capsules (three groups combined). Salmon, 2, 4 and 6 capsules provided 0·82, 0·24, 0·47 and 0·69 g/d of LC n-3 fatty acids. Salmon provided 7 µg/d and capsules < 0·02 µg/d of Se. The predictive model (r(2) 0·31, P = 0·001) showed that increases in erythrocyte LC n-3 levels were similar when intakes of 0·82 g/d LC n-3 from salmon or capsules (1·92 (95 % CI 1·35, 2·49) v. 2·32 (95 % 1·76, 2·88) %) were consumed. Plasma Se increased significantly more with salmon than with capsules (12·2 (95 % CI 6·18, 18·12) v. 1·57 (95 % CI - 2·32, 5·45) µg/l, P = 0·01). LC n-3 status was similarly improved with consumption of salmon and capsules, while consuming salmon had the added benefit of increasing Se status. This is of particular relevance to the New Zealand population that has marginal LC n-3 and Se status.


Assuntos
Ácidos Graxos Ômega-3/sangue , Óleos de Peixe/administração & dosagem , Salmão , Selênio/sangue , Adulto , Animais , Suplementos Nutricionais , Ingestão de Alimentos , Eritrócitos/metabolismo , Feminino , Humanos , Masculino , Nova Zelândia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA